Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
2.
Int J Nanomedicine ; 9: 3481-98, 2014.
Article in English | MEDLINE | ID: mdl-25092978

ABSTRACT

Therapeutic engineered nanoparticles (NPs), including ultrasmall superparamagnetic iron oxide (USPIO) NPs, may accumulate in the lower digestive tract following ingestion or injection. In order to evaluate the reaction of human colon cells to USPIO NPs, the effects of non-stabilized USPIO NPs (NS-USPIO NPs), oleic-acid-stabilized USPIO NPs (OA-USPIO NPs), and free oleic acid (OA) were compared in human HT29 and CaCo2 colon epithelial cancer cells. First the biophysical characteristics of NS-USPIO NPs and OA-USPIO NPs in water, in cell culture medium supplemented with fetal calf serum, and in cell culture medium preconditioned by HT29 and CaCo2 cells were determined. Then, stress responses of the cells were evaluated following exposure to NS-USPIO NPs, OA-USPIO NPs, and free OA. No modification of the cytoskeletal actin network was observed. Cell response to stress, including markers of apoptosis and DNA repair, oxidative stress and degradative/autophagic stress, induction of heat shock protein, or lipid metabolism was determined in cells exposed to the two NPs. Induction of an autophagic response was observed in the two cell lines for both NPs but not free OA, while the other stress responses were cell- and NP-specific. The formation of lipid vacuoles/droplets was demonstrated in HT29 and CaCo2 cells exposed to OA-USPIO NPs but not to NS-USPIO NPs, and to a much lower level in cells exposed to equimolar concentrations of free OA. Therefore, the induction of lipid vacuoles in colon cells exposed to OA utilized as a stabilizer for USPIO NPs is higly amplified compared to free OA, and is not observed in the absence of this lipid in NS-USPIO NPs.


Subject(s)
Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Oleic Acid/chemistry , Oleic Acid/toxicity , Vacuoles/drug effects , Apoptosis/drug effects , Caco-2 Cells , HT29 Cells , Heat-Shock Proteins/metabolism , Humans , Lipids , Oleic Acid/pharmacokinetics , Particle Size , Stress, Physiological/drug effects
3.
ACS Appl Mater Interfaces ; 5(9): 3581-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23578059

ABSTRACT

Nanoparticles (NPs) are being used or explored for the development of biomedical applications in diagnosis and therapy, including imaging and drug delivery. Therefore, reliable tools are needed to study the behavior of NPs in biological environment, in particular the transport of NPs across biological barriers, including the blood-brain tumor barrier (BBTB), a challenging question. Previous studies have addressed the translocation of NPs of various compositions across cell layers, mostly using only one type of cells. Using a coculture model of the human BBTB, consisting in human cerebral endothelial cells preloaded with ultrasmall superparamagnetic iron oxide nanoparticles (USPIO NPs) and unloaded human glioblastoma cells grown on each side of newly developed ultrathin permeable silicon nitride supports as a model of the human BBTB, we demonstrate for the first time the transfer of USPIO NPs from human brain-derived endothelial cells to glioblastoma cells. The reduced thickness of the permeable mechanical support compares better than commercially available polymeric supports to the thickness of the basement membrane of the cerebral vascular system. These results are the first report supporting the possibility that USPIO NPs could be directly transferred from endothelial cells to glioblastoma cells across a BBTB. Thus, the use of such ultrathin porous supports provides a new in vitro approach to study the delivery of nanotherapeutics to brain cancers. Our results also suggest a novel possibility for nanoparticles to deliver therapeutics to the brain using endothelial to neural cells transfer.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/cytology , Brain/metabolism , Endothelial Cells/metabolism , Ferrosoferric Oxide/pharmacokinetics , Glioblastoma/metabolism , Magnetite Nanoparticles/chemistry , Biological Transport , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , DNA/metabolism , Endothelial Cells/chemistry , Endothelial Cells/cytology , Ferrosoferric Oxide/administration & dosage , Ferrosoferric Oxide/chemistry , Glioblastoma/chemistry , Humans , Magnetite Nanoparticles/administration & dosage , Models, Biological , Silicon Compounds
4.
Cell Biol Toxicol ; 29(1): 39-58, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23161112

ABSTRACT

A route of accumulation and elimination of therapeutic engineered nanoparticles (NPs) may be the kidney. Therefore, the interactions of different solid-core inorganic NPs (titanium-, silica-, and iron oxide-based NPs) were studied in vitro with the MDCK and LLC-PK epithelial cells as representative cells of the renal epithelia. Following cell exposure to the NPs, observations include cytotoxicity for oleic acid-coated iron oxide NPs, the production of reactive oxygen species for titanium dioxide NPs, and cell depletion of thiols for uncoated iron oxide NPs, whereas for silica NPs an apparent rapid and short-lived increase of thiol levels in both cell lines was observed. Following cell exposure to metallic NPs, the expression of the tranferrin receptor/CD71 was decreased in both cells by iron oxide NPs, but only in MDCK cells by titanium dioxide NPs. The tight association, then subsequent release of NPs by MDCK and LLC-PK kidney epithelial cells, showed that following exposure to the NPs, only MDCK cells could release iron oxide NPs, whereas both cells released titanium dioxide NPs. No transfer of any solid-core NPs across the cell layers was observed.


Subject(s)
Epithelial Cells/drug effects , Ferric Compounds/toxicity , Kidney Tubules/drug effects , Metal Nanoparticles/toxicity , Oxidative Stress/drug effects , Silicon Dioxide/toxicity , Titanium/toxicity , Animals , Antigens, CD/metabolism , Cell Survival/drug effects , DNA Replication/drug effects , Dogs , Epithelial Cells/metabolism , Epithelial Cells/pathology , Ferric Compounds/metabolism , Kidney Tubules/metabolism , LLC-PK1 Cells , Madin Darby Canine Kidney Cells , Receptors, Transferrin/metabolism , Silicon Dioxide/metabolism , Swine , Titanium/metabolism
5.
J Med Chem ; 55(11): 4990-5002, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22533316

ABSTRACT

Ten oxidosqualene cyclase inhibitors with high efficacy as cholesterol-lowering agents and of different chemical structure classes were evaluated as potential anticancer agents against human cancer cells from various tissue origins and nontumoral human-brain-derived endothelial cells. Inhibition of cancer cell growth was demonstrated at micromolar concentrations, comparable to the concentrations of statins necessary for antitumor effect. Human glioblastoma cells were among the most sensitive cells. These compounds were also able to decrease the proliferation of angiogenic brain-derived endothelial cells, as a model of tumor-induced neovasculation. Additive effects in human glioblastoma cells were also demonstrated for oxidosqualene cyclase inhibitors in combination with atorvastatin while maintaining selectivity against endothelial cells. Thus, not only statins targeting the 3-hydroxy-3-methylglutaryl coenzyme A reductase but also inhibitors of oxidosqualene cyclase decrease tumor growth, suggesting new therapeutic opportunities of combined anti-cholesterol agents for dual treatment of glioblastoma.


Subject(s)
Alkynes/pharmacology , Anticholesteremic Agents/pharmacology , Antineoplastic Agents/pharmacology , Carbamates/pharmacology , Cyclohexanes/pharmacology , Heptanoic Acids/pharmacology , Intramolecular Transferases/antagonists & inhibitors , Pyrroles/pharmacology , Alkynes/chemical synthesis , Alkynes/chemistry , Angiogenesis Inhibitors/chemical synthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Atorvastatin , Brain/blood supply , Carbamates/chemical synthesis , Carbamates/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cricetinae , Cyclohexanes/chemical synthesis , Cyclohexanes/chemistry , Drug Screening Assays, Antitumor , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Microsomes, Liver/enzymology , Neovascularization, Pathologic/pathology , Structure-Activity Relationship
6.
Biochem J ; 441(3): 813-21, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22026563

ABSTRACT

Different types of NPs (nanoparticles) are currently under development for diagnostic and therapeutic applications in the biomedical field, yet our knowledge about their possible effects and fate in living cells is still limited. In the present study, we examined the cellular response of human brain-derived endothelial cells to NPs of different size and structure: uncoated and oleic acid-coated iron oxide NPs (8-9 nm core), fluorescent 25 and 50 nm silica NPs, TiO2 NPs (21 nm mean core diameter) and PLGA [poly(lactic-co-glycolic acid)]-PEO [poly(ethylene oxide)] polymeric NPs (150 nm). We evaluated their uptake by the cells, and their localization, generation of oxidative stress and DNA-damaging effects in exposed cells. We show that NPs are internalized by human brain-derived endothelial cells; however, the extent of their intracellular uptake is dependent on the characteristics of the NPs. After their uptake by human brain-derived endothelial cells NPs are transported into the lysosomes of these cells, where they enhance the activation of lysosomal proteases. In brain-derived endothelial cells, NPs induce the production of an oxidative stress after exposure to iron oxide and TiO2 NPs, which is correlated with an increase in DNA strand breaks and defensive mechanisms that ultimately induce an autophagy process in the cells.


Subject(s)
Autophagy/drug effects , Brain/cytology , Endothelial Cells/drug effects , Lysosomes/drug effects , Nanoparticles , Oxidative Stress/drug effects , Polymers/pharmacology , Autophagy/physiology , Brain/drug effects , Brain/metabolism , Cell Survival/drug effects , Cells, Cultured , DNA/biosynthesis , DNA Damage/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Lactic Acid/adverse effects , Lactic Acid/chemistry , Lactic Acid/pharmacology , Lysosomes/metabolism , Nanoparticles/adverse effects , Oxidative Stress/physiology , Polyglycolic Acid/adverse effects , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers/adverse effects , Titanium/adverse effects , Titanium/chemistry , Titanium/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
7.
Bioconjug Chem ; 22(7): 1422-32, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21668008

ABSTRACT

The chemical functionalization of cell-surface proteins of human primary fetal bone cells with hydrophilic bioorthogonal intermediates was investigated. Toward this goal, chemical pathways were developed for click reaction-mediated coupling of alkyne derivatives with cellular azido-expressing proteins. The incorporation via a tetraethylene glycol linker of a dipeptide and a reporter biotin allowed the proof of concept for the introduction of cell-specific peptide ligands and allowed us to follow the reaction in living cells. Tuning the conditions of the click reaction resulted in chemical functionalization of living human fetal osteoblasts with excellent cell survival.


Subject(s)
Alkynes/chemistry , Click Chemistry , Membrane Proteins/chemistry , Osteoblasts/cytology , Cell Membrane/chemistry , Cell Survival , Cells, Cultured , Fetus/cytology , Humans , Hydrophobic and Hydrophilic Interactions , Osteoblasts/chemistry , Tissue Engineering/methods
8.
ChemMedChem ; 2(12): 1799-806, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17918761

ABSTRACT

Ruthenium-arene complexes conjugated to ethacrynic acid were prepared as part of a strategy to develop novel glutathione-S-transferase (GST) inhibitors with alternate modes of activity through the organometallic fragment, ultimately to provide targeted ruthenium-based anticancer drugs. Enzyme kinetics and electrospray mass spectrometry experiments using GST P1-1 and its cysteine-modified mutant forms revealed that the complexes are effective enzyme inhibitors, but they also rapidly inactivate the enzyme by covalent binding at Cys 47 and, to a lesser extent, Cys 101. They are highly effective against the GST Pi-positive A2780 and A2780cisR ovarian carcinoma cell lines, are among the most effective ruthenium complexes reported so far, and target ubiquitous GST Pi overexpressed in many cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Organometallic Compounds/pharmacology , Ruthenium Compounds/pharmacology , Antineoplastic Agents/chemistry , Enzyme Inhibitors/chemistry , Kinetics , Magnetic Resonance Spectroscopy , Organometallic Compounds/chemistry , Ruthenium Compounds/chemistry , Spectrometry, Mass, Electrospray Ionization
9.
Chem Biol ; 12(8): 867-72, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16125098

ABSTRACT

The determination of enzyme activity or inhibition in intact living cells is a problem in the development of inhibitors for intracellular proteases. The production of fluorescent protoporphyrin IX (PpIX) from the nonfluorescent (N)-Gly/Pro-5-aminolevulinic acid (ALA) substrates was used to evaluate the prolyl/glycyl-specific dipeptidylpeptidase IV (DPPIV)-like and prolyloligopeptidase (POP)-like activities of human cells. The results demonstrated that whereas POP-like activity could be attributed to the actual POP, the DPPIV-like activity could be related to actual DPPIV only in one colon cell line. In the other breast and colon cell lines, DPPIV-like activity was intracellular and displayed by other prolyl-specific aminopeptidases. Our experiments also demonstrated the involvement of glycyl-specific proteases in the processing of ALA precursors. These observations have important consequences for the development and evaluation of selective inhibitors for these enzymes.


Subject(s)
Cysteine Endopeptidases/analysis , Fluorescent Dyes , Protoporphyrins/biosynthesis , Serine Endopeptidases/analysis , Adenosine Deaminase/metabolism , Aminolevulinic Acid/analogs & derivatives , Aminolevulinic Acid/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Dipeptidyl Peptidase 4/metabolism , Drug Design , Female , Glycoproteins/metabolism , Humans , Kinetics , Male , Methods , Prolyl Oligopeptidases , Protease Inhibitors/chemical synthesis , Protease Inhibitors/pharmacology , Protoporphyrins/analysis , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...